999 resultados para Nuclear Ribonucleoprotein B1


Relevância:

90.00% 90.00%

Publicador:

Resumo:

Heterogeneous nuclear ribonucleoprotein (hnRNP) A2 binds a 21-nucleotide myelin basic protein mRNA response element, the A2RE, and A2RE-like sequences in other localized mRNAs, and is a trans-acting factor in oligodendrocyte cytoplasmic RNA trafficking. Recombinant human hnRNPs A1 and A2 were used in a biosensor to explore interactions with A2RE and the cognate oligodeoxyribonucleotide. Both proteins have a single site that bound oligonucleotides with markedly different sequences but did not bind in the presence of heparin. Both also possess a second, specific site that bound only A2RE and was unaffected by heparin, hnRNP A2 bound A2RE in the latter site with a K-d near 50 nM, whereas the K-d for hnRNP A1 was above 10 muM. UV cross-linking assays led to a similar conclusion. Mutant A2RE sequences, that in earlier qualitative studies appeared not to bind hnRNP A2 or support RNA trafficking in oligodendrocytes, had dissociation constants above 5 muM for this protein. The two concatenated RNA recognition motifs (RRMs), but not the individual RRMs, mimicked the binding behavior of hnRNP A2. These data highlight the specificity of the interaction of A2RE with these hnRNPs and suggest that the sequence-specific A2RE-binding site on hnRNP A2 is formed by both RRMs acting in cis.

Relevância:

80.00% 80.00%

Publicador:

Resumo:

The removal of noncoding sequences, or introns, from the eukaryotic messenger RNA precursors is catalyzed by a ribonucleoprotein complex known as the spliceosome. In most eukaryotes, two distinct classes of introns exist, each removed by a specific type of spliceosome. The major, U2-type introns account for over 99 % of all introns, and are almost ubiquitous. The minor, U12-type introns are found in most but not all eukaryotes, and reside in conserved locations in a specific set of genes. Due to their slow excision rates, the U12-type introns are expected to be involved in the regulation of the genes containing them by inhibiting the maturation of the messenger RNAs. However, little information is currently available on how the activity of the U12-dependent spliceosome itself is regulated. The levels of many known splicing factors are regulated through unproductive alternative splicing events, which lead to inclusion of premature STOP codons, targeting the transcripts for destruction by the nonsense-mediated decay pathway. These alternative splice sites are typically found in highly conserved sequence elements, which also contain binding sites for factors regulating the activation of the splice sites. Often, the activation is achieved by binding of products of the gene in question, resulting in negative feedback loops. In this study, I show that U11-48K, a protein factor specific to the minor spliceosome, specifically recognizes the U12-type 5' splice site sequence, and is essential for proper function of the minor spliceosome. Furthermore, the expression of U11-48K is regulated through a feedback mechanism, which functions through conserved sequence elements that activate alternative splicing and nonsense-mediated decay. This mechanism is conserved from plants to animals, highlighting both the importance and early origin of this mechanism in regulating splicing factors. I also show that the feedback regulation of U11-48K is counteracted by a component of the major spliceosome, the U1 small nuclear ribonucleoprotein particle, as well as members of the hnRNP F/H protein family. These results thus suggest that the feedback mechanism is finely tuned by multiple factors to achieve precise control of the activity of the U12-dependent spliceosome.

Relevância:

80.00% 80.00%

Publicador:

Resumo:

Small nuclear ribonucleoprotein particles (snRNPs) and non-snRNP splicing factors containing a serine/arginine-rich domain (SR proteins) concentrate in 'speckles' in the nucleus of interphase cells(1). It is believed that nuclear speckles act as storage sites for splicing factors while splicing occurs on nascent transcripts(2). Splicing factors redistribute in response to transcription inhibition(3,4) or viral infection(5), and nuclear speckles break down and reform as cells progress through mitosis(6). We have now identified and cloned a kinase, SRPK1, which is regulated by the cell cycle and is specific for SR proteins; this kinase is related to a Caenorhabditis elegans kinase and to the fission yeast kinase Dsk1 (ref. 7). SRPK1 specifically induces the disassembly of nuclear speckles, and a high level of SRPK1 inhibits splicing in vitro. Our results indicate that SRPK1 mag have a central role in the regulatory network for splicing, controlling the intranuclear distribution of splicing factors in interphase cells, and the reorganization of nuclear speckles during mitosis.

Relevância:

80.00% 80.00%

Publicador:

Resumo:

Le diabète est une maladie chronique dont la principale caractéristique est un niveau plasmatique élevé de glucose, qui est causé soit par un défaut dans la production d’insuline, l’action de l’insuline, ou les deux à la fois. Plusieurs études ont démontré que l’hyperglycémie chronique peut mener à la dysfonction et même la défaillance de plusieurs organes, dont le coeur, le système vasculaire, les yeux et les reins, se traduisant par des infarctus du myocarde, des accidents cérébro-vasculaires et des complications rétinales et rénales, respectivement. La néphropathie diabétique (DN) est la principale cause de déficience rénale et affecte près de 25-40% des patients diabétiques. La DN est invariablement associée à un risque élevé d’accident cérébrovasculaire et de dysfonction cardivasculaire. L’angiotensinogène (Agt) est l’unique précurseur de tous les types d’angiotensines. En plus du système rénine-angiotensine (RAS) sytémique, le rein possède son propre système intrarénal et exprime tous les composants du RAS. L’Agt est fortement exprimé dans les cellules du tubule proximal rénal (RPTC) et y est converti en angiotensine II (AngII), le peptide biologiquement actif du RAS. Les patients diabétiques présentent de hauts niveaux d’AngII et une augmentation de l’expression des gènes du RAS, suggérant que l’activation du RAS intrarénal joue un rôle important dans la progression de la DN. Les mécanismes qui contrôlent la régulation du niveau rénal d’Agt par l’hyperglycémie et l’insuline demeurent mal compris. Le but global de cette thèse est de mieux comprendre les mécanismes moléculaires qui contrôlent l’expression du gène Agt chez la souris Akita (un modèle murin de diabète de type 1). Dans cette optique, la première partie de la thèse se concentre sur deux facteurs de transcription de la famille des ribonucléoprotéines nucléaires hétérogènes (hnRNP). Chan et collaborateurs ont déjà identifié 2 protéines nucléaires hnRNP F et hnRNP K, de 48kD et 70kD respectivement. HnRNP F et hnRNP K forment un hétérodimère et se lient à l’élément de réponse à l’insuline (IRE) présent dans le promoteur du gène Agt du rat et inhibent la transcription du gène Agt in vitro. Afin de déterminer si hnRNP F / K sont responsables de l’inhibition de l’expression rénale de Agt par l’insuline in vivo, nous avons étudié des souris Akita males traités ou non avec des implants d’insuline pour une période de 4 semaines. Des souris non-Akita males ont été employées comme contrôles. Les souris Akita développent de l’hypertension et de l’hypertrophie rénale. Le traitement à l’insuline rétablit les niveaux de glucose plasmatiques et la pression systolique (SBP), et atténue l’hypertrophie rénale, l’albuminurie (ratio albumine/créatinine urinaire, ACR) et les niveaux urinaires d’Agt et AngII chez les souris Akita. De plus, le traitement à l’insuline inhibe l’expression rénale du gène Agt, tout en augmentant l’expression des gènes hnRNP F, hnRNP K et ACE2 (enzyme de conversion de l’angiotensine-2). Dans des RPTC in vitro, l’insuline inhibe Agt, mais stimule l’expression de hnRNP F et hnRNP K en présence de hautes concentrations de glucose, et ce via la voie de signalisation MAPK p44/42 (protéine kinase activée par un mitogène). La transfection avec des petits ARN interférents (siRNA) contre hnRNP F et hnRNP K prévient l’inhibition de l’expression d’Agt par l’insuline dans les RPTC. Cette étude démontre bien que l’insuline prévient l’hypertension et atténue les dommages rénaux observés chez les souris Akita diabétiques, en partie grâce à la suppression de la transcription rénale de Agt, via une augmentation de l’expression de hnRNP F et hnRNP K. La seconde partie de cette thèse change de focus et se tourne vers le facteur Nrf2 (nuclear factor erythroid 2-related factor 2). Nrf2 est un facteur de transcription qui contrôle les gènes de la réponse antioxydante cellulaire en réponse au stress oxydant ou aux électrophiles. Le but de cette étude est d’examiner l’impact de la surexpression de la catalase (Cat) dans les RPTC sur l’expression du gène Agt via Nrf2 et sur le développement de l’hypertension et des dommages rénaux résultants chez les souris diabétiques Akita transgéniques (Tg). Nos études ont démontré que la surexpression de Cat dans les souris Akita Cat-Tg normalise la SBP, atténue les dommages rénaux et inhibe l’expression des gènes Nrf2 et Agt dans les RPTC. In vitro, le glucose élevé (HG) et l’oltipraz (un activateur de Nrf2) stimulent l’expression de Nrf2 et Agt, et cet effet peut être bloqué par la trigonelline (inhibiteur de Nrf2), des siRNA contre Nrf2, des antioxydants ou des inhibiteurs pharmacologiques NF-κB et MAPK p38. La suppression de sites de réponse à Nrf2 présents dans le promoteur du gène Agt du rat abolit la stimulation par l’oltipraz. Finalement, des souris males adultes non-transgéniques traitées avec l’oltipraz montrent une augmentation de l’expression de Nrf2 et Agt dans leurs RPTC et cette augmentation peut être normalisée par la trigonelline. Ces données permettent d’identifier un nouveau mécanisme d’action de Nrf2, par la stimulation du gène Agt intrarénal et l’activation du RAS, qui induisent l’hypertension et les dommages rénaux par le glucose élevé et les espèces réactives de l’oxygène chez les souris diabétiques. Nos conclusions permettent de démontrer que l’insuline induit l’expression de hnRNP F et hnRNP K, qui jouent ensuite un rôle protecteur en prévenant l’hypertension. La surexpression de la catalase dans les RPTC vient quant à elle atténuer l’activation de Nrf2 et ainsi réduit la SBP chez les souris Akita.

Relevância:

80.00% 80.00%

Publicador:

Resumo:

Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP)

Relevância:

80.00% 80.00%

Publicador:

Resumo:

Introduction Phospholipase Cb1 (PLC-β1) is a key player in the regulation of nuclear inositol lipid signaling and of a wide range of cellular functions, such as proliferation and differentiation (1,2,3). PLCb1 signaling depends on the cleavage of phosphatidylinositol 4,5-bisphosphate and the formation of the second messengers diacylglycerol and Inositol tris-phosphate which activate canonical protein kinase C (cPKC) isoforms. Here we describe a proteomic approach to find out a potential effector of nuclear PLC-b1 dependent signaling during insulin stimulated myogenic differentiation. Methods Nuclear lysates obtained from insulin induced C2C12 myoblasts were immunoprecipitated with anti-phospho-substrate cPKC antibody. Proteins, stained with Comassie blue, were excised, digested and subsequently analysed in LC-MS/MS. For peptide sequence searching, the mass spectra were processed and analyzed using the Mascot MS/MS ion search program with the NCBI database. Western blotting, GST-pull down and co-immunoprecipitation were performed to study the interaction between eEF1A2 and cPKCs. Site direct mutagenesis was performed to confirm the phosphorylated motif recognized by the antibody. Immunofluorescence analysis, GFP-tagged eEF1A2 vector and subcellular fractionation were performed to study nuclear localization and relative distribution of eEF1A2. Results We have previously shown that PLC-β1 is greatly increased at the nuclear level during insulin-induced myoblasts differentiation and that this nuclear localization is essential for induction of differentiation. Thus, nuclear proteins of insulin stimulated C2C12 myoblasts, were immunoprecipitated with an anti-phospho-substrate cPKC antibody. After Electrophoretic gel separation of proteins immunoprecipitated, several molecules were identified by LC-MS/MS. Among these most relevant and unexpected was eukaryotic elongation factor 1 alpha 2 (eEF1A2). We found that eEF1A2 is phosphorylated by PKCb1 and that these two molecules coimmunolocalized at the nucleolar level. eEF1A2 could be phosphorylated in many sites among which both threonine and serine residues. By site direct mutagenesis we demonstrated that it is the serine residue of the motif recognized by the antibody that is specifically phosphorylated by PKCb1. The silencing of PLCb1 gives rise to a reduction of expression and phosphorylation levels of eEF1A2 indicating this molecule as a target of nuclear PLCb1 regulatory network during myoblasts differentiation.

Relevância:

80.00% 80.00%

Publicador:

Resumo:

During central nervous system myelination, oligodendrocytes extend membrane processes towards an axonal contact site which is followed by ensheathment resulting in a compacted multilamellar myelin sheath. The formation of this axon-glial unit facilitates rapid saltatory propagation of action potentials along the axon and requires the synthesis and transport of copious amounts of lipids and proteins to the axon-glial contact site. Fyn is a member of the Src family of non receptor tyrosine kinases and inserted into the inner leaflet of the oligodendrocyte membrane by acylation. Fyn activity plays a pivotal role in the maturation of oligodendrocytes and the myelination process. It was suggested previously that Fyn kinase can be stimulated by binding of a neuronal ligand to oligodendroglial F3/ contactin, a glycosyl-phosphatidyl-inositol anchored immunoglobulin superfamily (IgSF) member protein. It could be shown here, that neuronal cell adhesion molecule L1 binds to oligodendrocytes in an F3-dependent manner and activates glial Fyn. In the search for downstream participants of this novel axon-glial signalling cascade, heterogeneous nuclear ribonucleoprotein (hnRNP) A2 was identified as a novel Fyn target in oligodendrocytes. HnRNP A2 was known to be involved in the localisation of translationally repressed myelin basic protein (MBP) mRNA by binding to a cis acting A2 response element (A2RE) present in the 3’ untranslated region. Transport of MBP mRNAs occurs in RNA-protein complexes termed RNA granules and translational repression during transport is achieved by hnRNP A2-mediated recruitment of hnRNP E1 to the granules. It could be shown here, that Fyn activity leads to enhanced translation of reporter mRNA containing a part of the 3’ UTR of MBP including the A2RE. Furthermore hnRNP E1 seems to dissociate from RNA granules in response to Fyn activity and L1 binding. These findings suggest a novel form of neuron- glial communication: Axonal L1 binding to oligodendroglial F3 activates Fyn kinase. Activated Fyn phosphorylates hnRNP A2 leading to removal of hnRNP E1 from RNA granules initiating the translation of MBP mRNA. MBP is the second most abundant myelin protein and mice lacking this protein show a severe hypomyelination phenotype. Moreover, the brains of Fyn knock out mice contain reduced MBP levels and are hypomyelinated. Hence, L1-mediated MBP synthesis via Fyn as a central molecule could be part of a regulatory mechanism required for myelinogenesis in the central nervous system.

Relevância:

80.00% 80.00%

Publicador:

Resumo:

In the central nervous system (CNS), oligodendrocytes form the multilamellar and compacted myelin sheath by spirally wrapping around defined axons with their specialised plasma membrane. Myelin is crucial for the rapid saltatory conduction of nerve impulses and for the preservation of axonal integrity. The absence of the major myelin component Myelin Basic Protein (MBP) results in an almost complete failure to form compact myelin in the CNS. The mRNA of MBP is sorted to cytoplasmic RNA granules and transported to the distal processes of oligodendrocytes in a translationally silent state. A main mediator of MBP mRNA localisation is the trans-acting factor heterogeneous nuclear ribonucleoprotein (hnRNP) A2 which binds to the cis-acting A2 response element (A2RE) in the 3’UTR of MBP mRNA. A signalling cascade had been identified that triggers local translation of MBP at the axon-glial contact site, involving the neuronal cell adhesion molecule (CAM) L1, the oligodendroglial plasma membrane-tethered Fyn kinase and Fyn-dependent phosphorylation of hnRNP A2. This model was confirmed here, showing that L1 stimulates Fyn-dependent phosphorylation of hnRNP A2 and a remodelling of A2-dependent RNA granule structures. Furthermore, the RNA helicase DDX5 was confirmed here acting together with hnRNP A2 in cytoplasmic RNA granules and is possibly involved in MBP mRNA granule dynamics.rnLack of non-receptor tyrosine kinase Fyn activity leads to reduced levels of MBP and hypomyelination in the forebrain. The multiadaptor protein p130Cas and the RNA-binding protein hnRNP F were verified here as additional targets of Fyn in oligodendrocytes. The findings point at roles of p130Cas in the regulation of Fyn-dependent process outgrowth and signalling cascades ensuring cell survival. HnRNP F was identified here as a novel constituent of oligodendroglial cytoplasmic RNA granules containing hnRNP A2 and MBP mRNA. Moreover, it was found that hnRNP F plays a role in the post-transcriptional regulation of MBP mRNA and that defined levels of hnRNP F are required to facilitate efficient synthesis of MBP. HnRNP F appears to be directly phosphorylated by Fyn kinase what presumably contributes to the initiation of translation of MBP mRNA at the plasma membrane.rnFyn kinase signalling thus affects many aspects of oligodendroglial physiology contributing to myelination. Post-transcriptional control of the synthesis of the essential myelin protein MBP by Fyn targets is particularly important. Deregulation of these Fyn-dependent pathways could thus negatively influence disorders involving the white matter of the nervous system.rnrn

Relevância:

80.00% 80.00%

Publicador:

Resumo:

Spinal muscular atrophy (SMA) is a lethal hereditary disease caused by homozygous deletion/inactivation of the survival of motoneuron 1 (SMN1) gene. The nearby SMN2 gene, despite its identical coding capacity, is only an incomplete substitute, because a single nucleotide difference impairs the inclusion of its seventh exon in the messenger RNA (mRNA). This splicing defect can be corrected (transiently) by specially designed oligonucleotides. Here we have developed a more permanent correction strategy based on bifunctional U7 small nuclear RNAs (snRNAs). These carry both an antisense sequence that allows specific binding to exon 7 and a splicing enhancer sequence that will improve the recognition of the targeted exon. When expression cassettes for these RNAs are stably introduced into cells, the U7 snRNAs become incorporated into small nuclear ribonucleoprotein (snRNP) particles that will induce a durable splicing correction. We have optimized this strategy to the point that virtually all SMN2 pre-mRNA becomes correctly spliced. In fibroblasts from an SMA patient, this approach induces a prolonged restoration of SMN protein and ensures its correct localization to discrete nuclear foci (gems).

Relevância:

80.00% 80.00%

Publicador:

Resumo:

The origin and structure of P55$\sp{\rm gag},$ a gag encoded polyprotein lacking the nucleocapsid protein, NCp10, have been explored. Evidence shows that P55$\sp{\rm gag}$ is formed by non-viral proteolytic cleavage of the Moloney murine leukemia virus (MoMuLV)gag precursor protein, Pr65$\sp{\rm gag}.$ P55$\sp{\rm gag}$ is produced in cells infected by a viral protease deletion mutant and by a recombinant murine sarcoma virus known to lack the protease gene, implying that a cellular protease is responsible for the cleavage. Structural and immunological studies show that the protein cleavage site is upstream of the CAp30-NCp10 viral proteolytic junction, implying that P55$\sp{\rm gag}$ lacks the carboxy-terminal residues of CAp30. During the course of studying P55$\sp{\rm gag},$ another protein was discovered, which I named nucleocapsid-related protein(NCRP). NCRP possesses the portion of CAp30 that is lacking in P55$\sp{\rm gag}.$ NCRP possesses antigenic epitopes present in CAp30 and NCp10. NCRP was observed in virus lysates and in nuclear lysates of MoMuLV infected cells; it was not detected in the cytoplasmic fractions of MoMuLV infected cells. Our results indicated that NCRP originates from Pr65$\sp{\rm gag},$ resulting from the same cellular proteolytic cleavage event that produces the viral cellular protein P55$\sp{\rm gag}.$ P55$\sp{\rm gag}$- and NCRP-like proteins also were observed in AKV murine leukemia virus (AKV MuLV) and feline leukemia virus (FeLV) infected cells and in their respective virus particles. The site of cleavage that yields P55$\sp{\rm gag}$ and NCRP is within the carboxy terminus of CAp30, likely within a motif highly conserved among mammalian type C retroviruses. This new motif, called the capsid conserved motif (CCM), overlaps a region containing both a possible bipartite nuclear targeting sequence and a region homologous with the U1 small nuclear ribonucleoprotein 70-kD protein. This domain, when intact, may act as a nuclear targeting sequence for the gag precursor proteins Pr65$\sp{\rm gag}$ and CAp30. Nuclei of cells infected with MoMuLV were examined for the presence of gag proteins. Both Pr65$\sp{\rm gag}$ and CAp30 were detected in the nuclear fraction of MoMuLV, AKV MuLV and FeLV infected cells. P55$\sp{\rm gag}$ was never detected in the nucleus of MoMuLV, AKV MuLV and FeLV infected cells or in their respective virus particles. I propose that NCRP may be involved in sequestering viral genomic RNA for the purposes of encapsidation and intracellular viral genomic RNA dimerization. ^

Relevância:

80.00% 80.00%

Publicador:

Resumo:

The 3' cleavage generating non-polyadenylated animal histone mRNAs depends on the base pairing between U7 snRNA and a conserved histone pre-mRNA downstream element. This interaction is enhanced by a 100 kDa zinc finger protein (ZFP100) that forms a bridge between an RNA hairpin element upstream of the processing site and the U7 small nuclear ribonucleoprotein (snRNP). The N-terminus of Lsm11, a U7-specific Sm-like protein, was shown to be crucial for histone RNA processing and to bind ZFP100. By further analysing these two functions of Lsm11, we find that Lsm11 and ZFP100 can undergo two interactions, i.e. between the Lsm11 N-terminus and the zinc finger repeats of ZFP100, and between the N-terminus of ZFP100 and the Sm domain of Lsm11, respectively. Both interactions are not specific for the two proteins in vitro, but the second interaction is sufficient for a specific recognition of the U7 snRNP by ZFP100 in cell extracts. Furthermore, clustered point mutations in three phylogenetically conserved regions of the Lsm11 N-terminus impair or abolish histone RNA processing. As these mutations have no effect on the two interactions with ZFP100, these protein regions must play other roles in histone RNA processing, e.g. by contacting the pre-mRNA or additional processing factors.

Relevância:

80.00% 80.00%

Publicador:

Resumo:

The U7 small nuclear ribonucleoprotein (U7 snRNP) is an essential factor mediating the unique 3’end processing of non-polyadenylated, replication-dependent histone mRNAs in metazoans. These histone genes expression and processing of their transcripts are cell cycle-regulated mechanisms that recruit a number of specific proteins as well as common factors required for expression and maturation of polyadenylated mRNAs. However, despite all the knowledge we have so far, there are still gaps in understanding of core histone RNA 3’ end processing, its coupling to transcription and regulation during cell cycle. To further elucidate this phenomena we used affinity chromatography based on tagged version of U7 snRNA molecule to identify proteins associated with U7 snRNP/U7 snRNA that could be potentially involved in core histone genes expression in human cells. Mass spectrometric analysis of affinity-purified fraction revealed, among others, multifunctional RNA/DNAbinding protein FUS/TLS (fused in sarcoma/translocated in liposarcoma) as a new factor interacting with U7 snRNA/RNP. Co-immunoprecipitation and RIP experiments confirmed the binding between FUS and the U7 RNA/snRNP. Interestingly, FUS:U7 snRNA interaction seems to be activated in S phase where the core histone genes are expressed. Moreover, FUS co-fractionates in 10-50% continuous glycerol gradient with other factors involved in histone premRNAs 3’end processing. However, this unique 3’end maturation was not disturbed upon FUS knockdown. Instead, we found that FUS depletion leads to a de-regulation of expression from selected histone promoters, suggesting that FUS is rather involved in regulation of core histone genes transcription. Thus, FUS bound to U7 snRNP can play a role in coupling between transcription and 3’end processing of replication dependant histone mRNAs.

Relevância:

80.00% 80.00%

Publicador:

Resumo:

In this study we demonstrate, at an ultrastructural level, the in situ distribution of heterogeneous nuclear RNA transcription sites after microinjection of 5-bromo-UTP (BrUTP) into the cytoplasm of living cells and subsequent postembedding immunoelectron microscopic visualization after different labeling periods. Moreover, immunocytochemical localization of several pre-mRNA transcription and processing factors has been carried out in the same cells. This high-resolution approach allowed us to reveal perichromatin regions as the most important sites of nucleoplasmic RNA transcription and the perichromatin fibrils (PFs) as in situ forms of nascent transcripts. Furthermore, we show that transcription takes place in a rather diffuse pattern, without notable local accumulation of transcription sites. RNA polymerase II, heterogeneous nuclear ribonucleoprotein (hnRNP) core proteins, general transcription factor TFIIH, poly(A) polymerase, splicing factor SC-35, and Sm complex of small nuclear ribonucleoproteins (snRNPs) are associated with PFs. This strongly supports the idea that PFs are also sites of major pre-mRNA processing events. The absence of nascent transcripts, RNA polymerase II, poly(A) polymerase, and hnRNPs within the clusters of interchromatin granules rules out the possibility that this domain plays a role in pre-mRNA transcription and polyadenylation; however, interchromatin granule-associated zones contain RNA polymerase II, TFIIH, and Sm complex of snRNPs and, after longer periods of BrUTP incubation, also Br-labeled RNA. Their role in nuclear functions still remains enigmatic. In the nucleolus, transcription sites occur in the dense fibrillar component. Our fine structural results show that PFs represent the major nucleoplasmic structural domain involved in active pre-mRNA transcriptional and processing events.

Relevância:

80.00% 80.00%

Publicador:

Resumo:

The stem-loop binding protein (SLBP1) binds the 3′ stem-loop of histone pre-mRNA and is required for efficient processing of histone transcripts in the nucleus. We examined the localization of SLBP1 in the germinal vesicle of Xenopus laevis oocytes. In spread preparations of germinal vesicle contents, an anti-SLBP1 antibody stained coiled bodies and specific chromosomal loci, including terminal granules, axial granules, and some loops. After injection of myc-tagged SLBP1 transcripts into the oocyte cytoplasm, newly translated myc-SLBP1 protein was detectable in coiled bodies within 4 h and in terminal and axial granules by 8 h. To identify the region(s) of SLBP1 necessary for subnuclear localization, we subcloned various parts of the SLBP1 cDNA and injected transcripts of these into the cytoplasm of oocytes. We determined that 113 amino acids at the carboxy terminus of SLBP1 are sufficient for coiled body localization and that disruption of a previously defined RNA-binding domain did not alter this localization. Coiled bodies also contain the U7 small nuclear ribonucleoprotein particle (snRNP), which participates in cleavage of the 3′ end of histone pre-mRNA. The colocalization of SLBP1 and the U7 snRNP in the coiled body suggests coordinated control of their functions, perhaps through a larger histone-processing particle. Some coiled bodies are attached to the lampbrush chromosomes at the histone gene loci, consistent with the view that coiled bodies in the oocyte recruit histone-processing factors to the sites of histone pre-mRNA transcription. The non-histone chromosomal sites at which SLBP1 is found include the genes coding for 5 S rRNA, U1 snRNA, and U2 snRNA, suggesting a wider role for SLBP1 in the biosynthesis of small non-spliced RNAs.

Relevância:

80.00% 80.00%

Publicador:

Resumo:

Exonic splicing enhancer (ESE) sequences are important for the recognition of splice sites in pre-mRNA. These sequences are bound by specific serine-arginine (SR) repeat proteins that promote the assembly of splicing complexes at adjacent splice sites. We have recently identified a splicing “coactivator,” SRm160/300, which contains SRm160 (the SR nuclear matrix protein of 160 kDa) and a 300-kDa nuclear matrix antigen. In the present study, we show that SRm160/300 is required for a purine-rich ESE to promote the splicing of a pre-mRNA derived from the Drosophila doublesex gene. The association of SRm160/300 and U2 small nuclear ribonucleoprotein particle (snRNP) with this pre-mRNA requires both U1 snRNP and factors bound to the ESE. Independently of pre-mRNA, SRm160/300 specifically interacts with U2 snRNP and with a human homolog of the Drosophila alternative splicing regulator Transformer 2, which binds to purine-rich ESEs. The results suggest a model for ESE function in which the SRm160/300 splicing coactivator promotes critical interactions between ESE-bound “activators” and the snRNP machinery of the spliceosome.